Melanotan-2 Manufacturing Profile: SPPS Synthesis, Purification & Quality Control Standards

Melanotan-2 Manufacturing Profile: Complete Production & Quality Control Standards

Melanotan-2 (MT-2) is a synthetic heptapeptide analog of alpha-melanocyte stimulating hormone (α-MSH) that requires specialized manufacturing protocols to ensure consistent quality for research and pharmaceutical applications. This manufacturing profile provides comprehensive technical specifications for solid-phase peptide synthesis (SPPS), purification methodologies, analytical quality control testing, batch release criteria, stability protocols, and storage requirements that meet current Good Manufacturing Practice (cGMP) standards for pharmaceutical-grade peptide production.

The manufacture of Melanotan-2 presents unique challenges related to sequence complexity, oxidative cyclization requirements, and stability considerations that distinguish it from linear peptide production. Successful commercial-scale synthesis demands precise control of coupling efficiency, racemization prevention during cyclization, systematic impurity profiling, and validated analytical methods to ensure batch-to-batch reproducibility and regulatory compliance.

1. Chemical Structure and Synthesis Overview

Melanotan-2 is characterized by the amino acid sequence Ac-Nle-cyclo[Asp-His-D-Phe-Arg-Trp-Lys]-NH₂, representing a cyclized heptapeptide with critical structural modifications including N-terminal acetylation, norleucine substitution at position 1, D-phenylalanine incorporation at position 4, and lactam bridge formation between Asp3 and Lys8 side chains. These structural features confer metabolic stability, receptor selectivity, and extended biological half-life compared to the endogenous α-MSH peptide.

The molecular formula is C₅₀H₆₉N₁₅O₉ with a molecular weight of 1024.2 g/mol (free base). The synthesis strategy employs Fmoc-based solid-phase peptide synthesis on acid-labile resin with systematic side-chain protection, followed by on-resin or solution-phase lactam cyclization and final cleavage under acidic conditions. The manufacturing process must address specific technical challenges including incomplete cyclization, epimerization at D-Phe, deletion sequences, oxidation of Trp and His residues, and aggregation during lyophilization.

Table 1: Melanotan-2 Molecular Specifications
Parameter Specification
Chemical Name Ac-Nle-cyclo[Asp-His-D-Phe-Arg-Trp-Lys]-NH₂
Molecular Formula C₅₀H₆₉N₁₅O₉
Molecular Weight 1024.2 g/mol (free base)
CAS Number 121062-08-6
Peptide Class Cyclic heptapeptide, α-MSH analog
Modifications N-acetylation, Nle¹, D-Phe⁴, lactam bridge (Asp³-Lys⁸)
Counter-ion (typical) Acetate or trifluoroacetate

2. Solid-Phase Peptide Synthesis Protocol

Manufacturing-scale synthesis of Melanotan-2 employs automated or semi-automated SPPS using Fmoc chemistry on Rink amide MBHA resin (substitution level 0.4-0.7 mmol/g) to provide the C-terminal amide functionality. The synthesis strategy proceeds from C-terminus to N-terminus with sequential coupling of protected amino acids: Fmoc-Lys(Mtt)-OH, Fmoc-Trp(Boc)-OH, Fmoc-Arg(Pbf)-OH, Fmoc-D-Phe-OH, Fmoc-His(Trt)-OH, Fmoc-Asp(OtBu)-OH, and Fmoc-Nle-OH, followed by acetylation with acetic anhydride. The Mtt (4-methyltrityl) protecting group on lysine permits selective deprotection for lactam formation while maintaining other side-chain protection.

Critical process parameters include coupling reagent selection, reaction time optimization, temperature control, and prevention of racemization. Standard coupling protocols employ HBTU/HOBt or HATU/HOAt activation systems with DIEA base in DMF solvent. Each coupling cycle is monitored by Kaiser test or chloranil test to confirm >99% coupling efficiency before proceeding. D-Phe incorporation requires careful selection of coupling conditions to minimize epimerization, typically employing shorter activation times and lower temperatures (0-5°C) compared to L-amino acids.

The cyclization step represents the most critical operation in MT-2 synthesis. Following selective removal of the Lys(Mtt) group with 1-2% TFA in DCM, the side-chain amine is activated for lactam formation with the aspartic acid carboxylate. On-resin cyclization is performed using PyBOP, HATU, or EDC/HOBt coupling reagents with extended reaction times (4-24 hours) under dilute conditions to favor intramolecular cyclization over intermolecular oligomerization. Cyclization efficiency is monitored by HPLC analysis of test cleavages, with optimization targeting >85% cyclization yield.

Table 2: SPPS Manufacturing Parameters for Melanotan-2
Process Step Reagents/Conditions Time Critical Parameters
Resin Loading Rink amide MBHA (0.4-0.7 mmol/g) - Substitution uniformity
Fmoc Deprotection 20% piperidine in DMF 2 × 5-10 min Complete removal, avoid aspartimide
Amino Acid Coupling 4-5 eq AA, HBTU/HOBt, DIEA in DMF 30-60 min >99% coupling efficiency
D-Phe Coupling 4-5 eq, HATU/HOAt, 0-5°C 60-90 min Minimize epimerization (<1%)
Mtt Deprotection 1-2% TFA in DCM Multiple 5 min cycles Selective Lys deprotection
Lactam Cyclization PyBOP/DIEA or HATU, dilute conditions 4-24 hours >85% cyclization efficiency
N-Acetylation Ac₂O/DIEA in DMF 30 min Complete capping
Final Cleavage TFA/TIS/H₂O (95:2.5:2.5) 2-3 hours Scavenger optimization

Following N-terminal acetylation, the completed peptide-resin is subjected to acidolytic cleavage using TFA cocktails containing scavengers (TIS, water, EDT, or thioanisole) to prevent oxidation and alkylation of sensitive residues. The cleavage cocktail composition is optimized to minimize Trp oxidation and His alkylation while ensuring complete removal of acid-labile protecting groups. The crude peptide is precipitated in cold diethyl ether, collected by centrifugation or filtration, washed repeatedly to remove scavengers and cleavage byproducts, and dried under vacuum or nitrogen stream before purification.

3. Purification Methodologies and Process Development

Crude Melanotan-2 typically exhibits 40-70% purity following SPPS and cleavage, containing deletion sequences, incomplete cyclization products, oxidized variants, and residual protecting group artifacts. Purification to pharmaceutical-grade specifications (>98% purity) requires multi-stage chromatographic separation employing preparative reversed-phase HPLC (RP-HPLC) as the primary technique, with optional ion-exchange or size-exclusion chromatography for specific impurity removal or buffer exchange.

Preparative RP-HPLC is performed on C18 silica columns (particle size 5-10 μm, pore size 100-300 Å) using acetonitrile/water gradients with 0.1% TFA modifier. The separation strategy is developed through analytical method optimization, evaluating gradient slope, flow rate, column temperature, and mobile phase pH to achieve baseline resolution between MT-2 and critical impurities including linear precursor, partially cyclized intermediates, and stereoisomers. Typical gradient conditions employ 20-50% acetonitrile over 30-60 minutes at flow rates of 10-100 mL/min depending on column diameter and loading capacity.

Process-scale purification addresses loading capacity, recovery optimization, and solvent economy while maintaining resolution performance. Column loading is optimized at 5-20 mg crude peptide per mL column volume, with overloading causing peak broadening and reduced separation efficiency. Fractionation is performed using online UV detection at 220 nm and 280 nm, with fraction collection based on retention time windows and subsequent analytical HPLC verification of purity before pooling. Pooled fractions are concentrated by rotary evaporation, lyophilized to dryness, and subjected to analytical release testing.

Table 3: Preparative HPLC Purification Parameters
Parameter Specification Purpose/Notes
Column Type C18 reversed-phase, 100-300 Å pore Hydrophobic interaction separation
Particle Size 5-10 μm Balance pressure/resolution
Column Dimensions 21.2-50 mm ID × 150-250 mm L Scale-dependent selection
Mobile Phase A 0.1% TFA in water Ion-pairing agent
Mobile Phase B 0.1% TFA in acetonitrile Organic modifier
Gradient Profile 20-50% B over 30-60 min Optimized for impurity separation
Flow Rate 10-100 mL/min (scale-dependent) Column diameter optimization
Column Temperature Ambient to 40°C Resolution enhancement
Detection Wavelength 220 nm (primary), 280 nm (secondary) Peptide bond and aromatic detection
Loading Capacity 5-20 mg crude/mL column volume Maintain resolution efficiency
Injection Volume 5-20% column volume Band broadening minimization
Recovery Target >60% (pure product from crude) Economic viability threshold

Alternative purification strategies may be employed for specific applications or impurity profiles. Ion-exchange chromatography (IEX) on strong cation exchange (SCX) resins can effectively separate MT-2 from deletion sequences or uncyclized variants based on charge differences. Size-exclusion chromatography (SEC) provides desalting and removal of aggregates or polymer contaminants. For TFA-sensitive applications, TFA counter-ions are exchanged through lyophilization from dilute HCl or acetic acid solutions, or by passage through ion-exchange resins in acetate or chloride form.

Purification process validation includes demonstration of reproducibility across multiple batches, impurity clearance capability, recovery consistency, and absence of column degradation or carryover. Process analytical technology (PAT) approaches employ online UV spectroscopy, conductivity monitoring, and pH measurement to enable real-time process adjustment and quality-by-design manufacturing strategies compliant with ICH Q8-Q11 guidelines.

4. Analytical Quality Control Testing

Pharmaceutical-grade Melanotan-2 requires comprehensive analytical characterization employing orthogonal techniques to verify identity, purity, potency, and absence of contaminants. The analytical control strategy is designed according to ICH Q6B specifications for biotechnological/biological products, adapted for synthetic peptides with consideration of sequence-specific degradation pathways and manufacturing-related impurities.

Primary analytical methods include:

4.1 Identity Testing

High-Performance Liquid Chromatography (HPLC): Identity confirmation by retention time comparison to reference standard under defined analytical conditions (±2% tolerance). The method employs C18 column (4.6 × 150-250 mm, 5 μm particle size) with acetonitrile/water/0.1% TFA gradient, detecting at 220 nm. Retention time reproducibility requires temperature control (±2°C) and mobile phase preparation standardization.

Mass Spectrometry (MS): Molecular weight confirmation by electrospray ionization (ESI-MS) or matrix-assisted laser desorption ionization time-of-flight (MALDI-TOF-MS). The observed molecular weight should match theoretical value ±1 Da for the predominant counter-ion form (acetate or TFA salt). High-resolution MS (HR-MS) provides elemental composition verification with mass accuracy <5 ppm.

Amino Acid Analysis (AAA): Quantitative amino acid composition following acid or enzymatic hydrolysis confirms sequence identity. The method requires careful protocol selection to minimize losses of acid-labile residues (Trp) while ensuring complete hydrolysis. Results are compared to theoretical composition with acceptance criteria typically ±10% for most residues, ±20% for Trp.

4.2 Purity and Impurity Profiling

Analytical RP-HPLC: Primary purity assay employing the same chromatographic conditions as identity testing but with integration of all peaks >0.05% area. Pharmaceutical-grade specifications typically require >98.0% purity by area normalization, with individual impurity limits <1.0% and total impurities <2.0%. The method validation includes specificity demonstration, linearity (R² >0.999), accuracy (95-105% recovery), precision (RSD <2.0%), and robustness testing.

Peptide Content Determination: Absolute peptide content (w/w%) is determined by amino acid analysis, quantitative HPLC with external standardization, or nitrogen determination. This assay distinguishes peptide mass from counter-ions, residual solvents, and water content, typically specifying >80% peptide content on an as-is basis or >95% on a dried basis.

Related Substances: Systematic impurity identification employs LC-MS/MS analysis with fragmentation pattern interpretation. Common manufacturing-related impurities include deletion sequences (n-1, n-2 peptides), linear uncyclized precursor, incompletely deprotected variants, oxidized Met analogs (if present in sequence), and stereoisomers from D-Phe epimerization. Each identified impurity is monitored and controlled according to ICH Q3B guidelines, with qualification thresholds based on maximum daily dose.

4.3 Physical-Chemical Characterization

Appearance: Visual inspection confirms white to off-white lyophilized powder with no discoloration, foreign particles, or cake collapse. Color assessment uses standardized reference materials or spectrophotometric methods.

Water Content: Karl Fischer titration measures residual moisture, with specifications typically <5.0% w/w for lyophilized material to ensure stability and accurate dosing. Thermogravimetric analysis (TGA) provides alternative quantification with thermal profile information.

Residual Solvents: Gas chromatography (GC-FID or GC-MS) quantifies residual manufacturing solvents according to ICH Q3C guidelines. Class 2 solvents (acetonitrile, DCM, DMF) are limited to <410, 600, and 880 ppm respectively, while Class 3 solvents (ethanol, acetone, ethyl acetate) are limited to <5000 ppm.

Counter-Ion Analysis: Ion chromatography or potentiometric titration quantifies TFA or acetate counter-ions. TFA content typically ranges from 1-3 molar equivalents (10-20% w/w), affecting hygroscopicity, solubility, and formulation pH.

Table 4: Analytical QC Testing Panel for Melanotan-2
Test Method Specification
Appearance Visual inspection White to off-white powder
Identity (RT) RP-HPLC Conforms to reference ±2%
Identity (MW) ESI-MS or MALDI-TOF 1024.2 ±1 Da (free base)
Amino Acid Composition AAA (acid hydrolysis) Conforms to theory ±10-20%
Purity (HPLC) RP-HPLC (area %) >98.0%
Single Impurity RP-HPLC <1.0%
Total Impurities RP-HPLC <2.0%
Peptide Content AAA or qHPLC >80% (as-is) or >95% (dry basis)
Water Content Karl Fischer <5.0%
Residual TFA Ion chromatography Report value (typically 10-20%)
Residual Solvents GC-FID/MS Per ICH Q3C limits
Bacterial Endotoxins LAL assay <5 EU/mg
Bioburden Microbial enumeration <10 CFU/g (if applicable)

4.4 Microbiological Testing

For pharmaceutical applications, bacterial endotoxin testing is performed using Limulus Amebocyte Lysate (LAL) assay with specification <5 EU/mg for parenteral products. Non-sterile peptide materials may require bioburden enumeration (total aerobic count <10 CFU/g) and absence of specific objectionable organisms (E. coli, Salmonella, Pseudomonas aeruginosa, Staphylococcus aureus). Sterile formulations undergo sterility testing by direct inoculation or membrane filtration methods according to USP <71> or Ph. Eur. 2.6.1.

5. Batch Manufacturing Specifications and Release Criteria

Commercial manufacturing of Melanotan-2 employs batch production methodology with defined batch sizes ranging from 1-100 grams depending on market demand and equipment capacity. Each batch is assigned a unique lot number enabling traceability through raw material sourcing, synthesis operations, purification processing, analytical testing, and final packaging. Batch records document all manufacturing operations, in-process controls, deviations, and corrective actions according to cGMP requirements outlined in 21 CFR Part 211 and ICH Q7 guidelines.

In-process controls monitor critical quality attributes during synthesis and purification to enable real-time decision making and reduce batch failure risk. Key in-process tests include:

  • Coupling efficiency monitoring: Kaiser or chloranil testing after each amino acid addition ensures >99% coupling before proceeding to prevent deletion sequences
  • Test cleavage analysis: Small-scale analytical cleavage and HPLC analysis before full-scale cleavage confirms synthesis success and cyclization efficiency
  • Crude purity assessment: HPLC analysis of crude material predicts purification yield and identifies process improvements
  • Purification fraction analysis: Real-time HPLC testing of collected fractions ensures only specification-meeting material is pooled
  • Lyophilization monitoring: Temperature and vacuum profiling confirms complete drying and cake integrity

Batch release requires satisfactory completion of all analytical testing with results meeting pre-defined acceptance criteria. The quality control testing panel must demonstrate compliance with identity, purity, potency, and safety specifications before distribution authorization. Any out-of-specification (OOS) result triggers investigation according to validated procedures, with root cause analysis, impact assessment, and corrective/preventive action (CAPA) implementation.

Table 5: Batch Release Specifications - Pharmaceutical Grade
Parameter Test Method Acceptance Criteria
Appearance Visual White to off-white lyophilized powder, no foreign matter
Identification (HPLC) RP-HPLC retention time Conforms to reference standard ±2%
Identification (MS) ESI-MS Molecular weight 1024.2 ±1 Da (free base)
Sequence Confirmation Amino acid analysis Conforms to Ac-Nle-c[Asp-His-D-Phe-Arg-Trp-Lys]-NH₂
Purity by HPLC RP-HPLC (UV 220nm) >98.0% (area normalization)
Single Largest Impurity RP-HPLC <1.0%
Total Impurities RP-HPLC <2.0%
Peptide Content (dry basis) Quantitative AAA or HPLC >95.0%
Water Content Karl Fischer titration <5.0%
Acetonitrile (residual) GC-FID <410 ppm
DMF (residual) GC-FID <880 ppm
TFA Content Ion chromatography Report value
Bacterial Endotoxins LAL kinetic chromogenic <5.0 EU/mg
Heavy Metals ICP-MS <10 ppm total

Research-grade material may employ relaxed specifications (>95% purity, <5% total impurities) for cost optimization, while pharmaceutical-grade material for clinical trials or commercial distribution demands stringent control (>98% purity, <2% total impurities, comprehensive impurity characterization). The specification tier is selected based on intended use, regulatory requirements, and risk assessment considering patient safety and data quality objectives.

6. Stability Studies and Degradation Pathways

Melanotan-2 stability is governed by multiple degradation pathways including peptide bond hydrolysis, deamidation, oxidation, epimerization, and aggregation. Systematic stability studies under defined storage conditions establish shelf-life specifications, identify optimal packaging and storage requirements, and support regulatory submissions. Stability protocols follow ICH Q1A-Q1F guidelines adapted for peptide pharmaceuticals, employing validated stability-indicating analytical methods capable of detecting and quantifying degradation products.

The primary degradation pathways identified through forced degradation studies include:

  • Oxidation: Tryptophan and histidine residues are susceptible to oxidation under aerobic conditions, light exposure, and elevated pH, forming N-formylkynurenine, kynurenine, and histidine oxidation products detectable by UV absorption changes and LC-MS
  • Deamidation: Asparagine deamidation (if present in variants) and aspartic acid isomerization occur at elevated pH and temperature, creating isoaspartyl isomers with altered charge and retention time
  • Hydrolysis: Peptide bond cleavage at Asp-Pro, Asp-Gly, or acid-labile sites occurs under acidic or basic conditions, generating truncated fragments
  • Epimerization: D-Phe racemization to L-Phe forms diastereomeric impurities with reduced biological activity, accelerated by base exposure and elevated temperature
  • Aggregation: Intermolecular association through hydrophobic interactions, disulfide cross-linking, or β-sheet formation produces high molecular weight species detectable by SEC-HPLC

Accelerated stability studies at 40°C/75% RH for 6 months and long-term studies at 25°C/60% RH for 12-36 months monitor purity, potency, water content, and appearance. Stressed condition testing at elevated temperature (60°C), humidity (>90% RH), light exposure (1.2 million lux-hours), and pH extremes (pH 2-10) identifies worst-case degradation pathways and validates analytical method specificity. Stability sample testing employs the same analytical methods as release testing, with trending analysis to detect significant changes from initial values.

Table 6: Stability Storage Conditions and Testing Schedule
Study Type Condition Duration Testing Frequency
Long-term 25°C ± 2°C / 60% ± 5% RH 36 months 0, 3, 6, 9, 12, 18, 24, 36 months
Intermediate 30°C ± 2°C / 65% ± 5% RH 12 months 0, 6, 12 months
Accelerated 40°C ± 2°C / 75% ± 5% RH 6 months 0, 1, 2, 3, 6 months
Refrigerated (long-term) 5°C ± 3°C 36 months 0, 3, 6, 9, 12, 18, 24, 36 months
Frozen -20°C ± 5°C 36 months 0, 12, 24, 36 months
Freeze-thaw cycling -20°C to 25°C (3 cycles) - After each cycle
Photostability ICH Q1B conditions (1.2M lux-hr) - At completion

Stability data for lyophilized Melanotan-2 (TFA salt) demonstrate acceptable stability at refrigerated storage (2-8°C) with <2% purity loss over 24 months when stored in sealed containers with desiccant protection. Accelerated conditions (40°C/75% RH) show 3-5% purity reduction over 6 months, primarily due to Trp oxidation and deamidation. Photostability testing reveals significant photodegradation upon light exposure, mandating amber glass vials or light-protective secondary packaging for light-sensitive formulations.

Solution stability is significantly reduced compared to solid-state stability due to hydrolysis, oxidation, and aggregation in aqueous environment. Reconstituted solutions in sterile water or buffered saline demonstrate 85-95% potency retention at 2-8°C for 7-14 days, with pH-dependent stability (optimal pH 4-6) and concentration-dependent aggregation (increased at >5 mg/mL). For multi-dose applications, antimicrobial preservatives (benzyl alcohol 0.9%, metacresol 0.3%) are incorporated with compatibility verification through forced degradation studies.

7. Storage Requirements and Packaging Specifications

Proper storage and packaging are critical for maintaining Melanotan-2 quality throughout the product lifecycle from manufacturing through distribution to end-use. Storage condition specifications are established based on stability study results, packaging material compatibility testing, and regulatory requirements for peptide pharmaceuticals. The storage strategy addresses multiple degradation vectors including temperature, humidity, oxygen, light, and mechanical stress.

Recommended storage conditions for lyophilized Melanotan-2:

  • Temperature: Refrigerated storage at 2-8°C provides optimal stability for long-term storage (24-36 month shelf life). Frozen storage at -20°C extends stability beyond 36 months for bulk material. Room temperature storage (20-25°C) is acceptable for short-term distribution (3-6 months) with appropriate packaging protection.
  • Humidity: Lyophilized material is hygroscopic and requires moisture protection to prevent water uptake, cake collapse, and accelerated degradation. Sealed containers with desiccant pouches (silica gel or molecular sieve) maintain <5% moisture content during storage.
  • Light Protection: Amber glass vials or aluminum foil overwrap protect photolabile residues (Trp, His) from light-induced oxidation. UV-protective secondary packaging is required for transparent primary containers.
  • Oxygen Protection: Inert atmosphere packaging (nitrogen or argon purging) minimizes oxidative degradation during long-term storage. Oxygen-scavenging sachets provide additional protection for extended shelf-life requirements.
Table 7: Packaging and Storage Specifications
Component Specification Purpose
Primary Container Amber glass vial (Type I borosilicate) Light protection, chemical inertness
Closure Butyl rubber stopper, aluminum crimp seal Hermetic seal, moisture barrier
Fill Volume 2-20 mL vial (per dose size) Minimize headspace oxidation
Desiccant Silica gel (2-5g) or molecular sieve Humidity control <20% RH
Atmosphere Nitrogen or argon purge Oxygen exclusion (<2% O₂)
Secondary Packaging Cardboard carton with product insert Physical protection, labeling
Storage Temperature 2-8°C (refrigerated) Optimal stability (24-36 mo shelf life)
Alternative Storage -20°C (frozen) Extended stability (>36 months)
Shipping Conditions 2-8°C with temperature monitoring Cold chain maintenance

Primary packaging employs Type I borosilicate glass vials (USP/Ph. Eur. compliant) with dimensions matched to fill volume (2-20 mL) to minimize headspace and reduce oxidation potential. Butyl rubber stoppers meet USP Class VI requirements for biocompatibility and extractables/leachables profiles, with siliconization to facilitate needle penetration for reconstitution. Aluminum crimp seals provide tamper-evidence and hermetic sealing to maintain inert atmosphere throughout shelf life.

Packaging material qualification includes extractables/leachables testing according to USP <661> and <1663>, compatibility studies under accelerated and long-term conditions, and functional performance testing (seal integrity, moisture vapor transmission rate, oxygen permeability). Container closure integrity testing employs vacuum decay, helium leak detection, or dye ingress methods to verify hermetic seal maintenance.

Shipping and distribution require cold chain management with temperature monitoring devices (data loggers) to document compliance with 2-8°C specification during transit. Insulated shipping containers with refrigerant packs (gel packs or dry ice for frozen shipments) maintain temperature control for 24-96 hours depending on configuration. Qualification studies validate shipping container performance under worst-case seasonal and geographic conditions.

8. Certificate of Analysis (CoA) Template and Documentation

Each manufactured batch of Melanotan-2 is accompanied by a Certificate of Analysis (CoA) documenting all quality control testing results, manufacturing date, expiration date, storage conditions, and batch-specific information. The CoA serves as the primary quality documentation for customers, regulatory authorities, and internal quality assurance, providing objective evidence of specification compliance and manufacturing control.

Standard CoA format includes:

8.1 Product Identification Section

  • Product Name: Melanotan-2 (MT-2)
  • Chemical Name: Ac-Nle-cyclo[Asp-His-D-Phe-Arg-Trp-Lys]-NH₂
  • CAS Number: 121062-08-6
  • Molecular Formula: C₅₀H₆₉N₁₅O₉
  • Molecular Weight: 1024.2 g/mol (free base)
  • Batch/Lot Number: [Unique identifier]
  • Manufacture Date: [ISO 8601 format]
  • Retest/Expiration Date: [Based on stability data]
  • Quantity: [Mass in mg or g]
  • Grade: [Research/Pharmaceutical/Custom]

8.2 Analytical Testing Results

Table 8: Example Certificate of Analysis - Batch MT2-2024-1234
Test Parameter Method Specification Result
Appearance Visual White to off-white powder White powder
Identification (HPLC RT) RP-HPLC Conforms ±2% Conforms (12.34 min)
Identification (MS) ESI-MS 1024.2 ±1 Da 1024.5 Da
Purity (HPLC) RP-HPLC (220nm) >98.0% 98.7%
Single Impurity (max) RP-HPLC <1.0% 0.4%
Total Impurities RP-HPLC <2.0% 1.3%
Peptide Content (dry) AAA >95.0% 97.2%
Water Content Karl Fischer <5.0% 3.1%
TFA Content Ion chromatography Report 15.3% (w/w)
Acetonitrile (residual) GC-FID <410 ppm 22 ppm
Bacterial Endotoxins LAL kinetic <5.0 EU/mg <0.5 EU/mg

8.3 Storage and Handling Information

  • Storage Temperature: Store at 2-8°C in original container
  • Storage Conditions: Protect from light and moisture
  • Shelf Life: 24 months from manufacture date when stored as recommended
  • Reconstitution: Dissolve in sterile water or bacteriostatic water; use within 14 days when refrigerated
  • Handling Precautions: Use appropriate PPE; avoid inhalation and skin contact

8.4 Quality Assurance Statements

The CoA includes quality assurance declarations confirming:

  • Manufacturing compliance with cGMP requirements (21 CFR 211, ICH Q7)
  • Testing performance by qualified personnel using validated methods
  • Results representing the batch tested and meeting all specifications
  • Traceability to reference standards and calibration records
  • Authorized release by Quality Assurance with signature and date

8.5 Regulatory and Safety Information

  • Intended Use: For research purposes only / For manufacturing use
  • Safety Data Sheet: Reference to SDS document number
  • Regulatory Status: Research chemical / Drug substance / Custom designation
  • Country of Origin: [Manufacturing location]
  • Harmonized Tariff Code: [For import/export]

CoA documentation is maintained in quality management systems with electronic archiving for retrieval, trend analysis, and regulatory inspection support. Digital CoAs with QR codes or verification links enable authentication and prevent counterfeiting. Batch genealogy records link CoA data to raw material certifications, equipment logs, environmental monitoring, and deviation investigations for complete traceability.

9. Regulatory Considerations and Manufacturing Standards

Melanotan-2 manufacturing for pharmaceutical applications requires compliance with region-specific regulations governing drug substance production, quality systems, and documentation practices. The regulatory framework varies by intended use (research, clinical trials, commercial distribution) and geographic market (US FDA, EMA, Health Canada, TGA, etc.), necessitating regulatory strategy development during process design and validation phases.

Key regulatory guidelines applicable to Melanotan-2 manufacturing include:

  • ICH Q7 (Good Manufacturing Practice for Active Pharmaceutical Ingredients): Establishes quality management system requirements, personnel qualifications, building and facility design, equipment qualification, raw material controls, production operations, validation, change control, and complaint handling procedures applicable to API manufacturing.
  • ICH Q6B (Specifications for Biotechnological/Biological Products): Provides guidance on analytical testing strategies, specification setting, and control approaches for peptide pharmaceuticals, adapted from recombinant protein guidelines.
  • ICH Q1A-Q1F (Stability Testing Guidelines): Defines stability study protocols, testing frequency, storage conditions, and data evaluation requirements supporting shelf-life determination and retest dating.
  • USP General Chapters: Monographs <621> (Chromatography), <1225> (Validation of Compendial Procedures), <1207> (Package Integrity Evaluation), and others provide standardized testing methods and acceptance criteria.
  • 21 CFR Part 211 (cGMP for Finished Pharmaceuticals): While focused on finished dosage forms, relevant sections apply to peptide processing including equipment cleaning validation, environmental monitoring, and quality control laboratory operations.

Manufacturing facility design incorporates classified areas (ISO 7/8 cleanrooms) for handling, weighing, and final packaging operations to minimize bioburden and particulate contamination. Environmental monitoring programs track viable and non-viable particles, surface contamination, and personnel hygiene compliance. Equipment qualification follows IQ/OQ/PQ protocols with documented performance verification and calibration maintenance.

Process validation employs lifecycle approach per ICH Q8-Q11, encompassing process design (stage 1), process qualification through prospective validation studies (stage 2), and continued process verification via statistical process control and ongoing trending (stage 3). Critical process parameters identified through risk assessment and design of experiments are monitored with proven acceptable ranges documented in batch records.

For clinical trial material production, additional considerations include investigational new drug (IND) application support, clinical batch record documentation, pharmacopeia compliance (where applicable), and coordination with clinical manufacturing and controls sections. Commercial manufacturing requires drug master file (DMF) preparation containing comprehensive chemistry, manufacturing, and controls (CMC) information for regulatory submission by drug product sponsors.

10. Troubleshooting and Process Optimization

Manufacturing challenges in Melanotan-2 production require systematic troubleshooting approaches combining analytical characterization, process parameter evaluation, and mechanistic understanding of synthetic chemistry and peptide behavior. Common issues and mitigation strategies include:

10.1 Low Crude Purity After Synthesis

Symptoms: HPLC analysis showing <40% main peak, multiple significant impurities, low overall recovery after cleavage.

Causes and Solutions:

  • Incomplete couplings: Verify Kaiser/chloranil test procedures, extend coupling times, increase amino acid excess, employ stronger activating reagents (HATU vs HBTU), use higher quality Fmoc-amino acids
  • Aspartimide formation: Reduce piperidine exposure time during Fmoc removal following Asp coupling, incorporate 0.1M HOBt in deprotection solution, consider alternative base (piperazine)
  • Incomplete cyclization: Optimize cyclization reagent (test PyBOP, HATU, EDC/HOBt), extend reaction time to 12-24 hours, reduce resin loading to favor intramolecular reaction, perform test cleavages to monitor progress
  • Epimerization: Minimize base exposure after D-Phe incorporation, use cold coupling conditions (0-5°C) for D-amino acids, reduce activation time before coupling

10.2 Poor Resolution During Purification

Symptoms: Co-elution of product with impurities, inability to achieve >98% purity, poor recovery due to broad fractionation.

Causes and Solutions:

  • Suboptimal gradient: Develop shallower gradient (15-35% vs 20-50% ACN), extend run time to improve resolution, optimize gradient shape (linear vs curved)
  • Column degradation: Monitor column performance with system suitability standards, implement column regeneration protocols, replace when efficiency drops >20%
  • Sample overload: Reduce injection mass to 5-10 mg/mL CV, optimize injection volume (<10% CV), consider larger diameter columns
  • Temperature effects: Control column temperature (±1°C), test 25-40°C range for improved selectivity
  • Mobile phase pH: Test formic acid (0.1%) vs TFA (0.1%) as ion-pair modifier, evaluate pH 2.0-3.0 range

10.3 Oxidation During Processing or Storage

Symptoms: Trp or His oxidation peaks in HPLC, UV absorption changes at 280nm, purity loss during stability studies.

Causes and Solutions:

  • Cleavage cocktail optimization: Increase EDT or thioanisole scavenger concentration (5-10%), minimize cleavage time to 2 hours, use fresh TFA and scavengers
  • Purification solvent quality: Use HPLC-grade acetonitrile and water, degas solvents with helium sparging, add 0.01% ascorbic acid as antioxidant
  • Lyophilization conditions: Minimize exposure to room air during transfer, use nitrogen or argon purge in lyophilizer, reduce sublimation temperature
  • Storage improvements: Package under inert atmosphere, add oxygen scavengers to vials, implement refrigerated or frozen storage, use amber glass for light protection

10.4 Aggregation or Precipitation

Symptoms: Turbid solutions upon reconstitution, high molecular weight peaks in SEC, reduced recovery, cake collapse during lyophilization.

Causes and Solutions:

  • Formulation optimization: Add excipients (mannitol 1-5%, sucrose 2-5%) as bulking agents, adjust pH to 4.5-5.5 optimal range, reduce peptide concentration <10 mg/mL
  • Lyophilization cycle development: Optimize freezing rate, adjust primary drying temperature to -25 to -35°C, extend secondary drying for residual moisture removal, perform thermal analysis (DSC) to determine collapse temperature
  • Reconstitution protocols: Use sterile water vs PBS (ionic strength effects), add solvent slowly with gentle mixing, avoid vigorous shaking which induces foaming

10.5 Batch-to-Batch Variability

Symptoms: Inconsistent crude purity (±10%), variable purification yield, analytical result trends outside normal ranges.

Causes and Solutions:

  • Raw material variability: Implement vendor qualification, test Fmoc-AA purity by HPLC, standardize resin batches, create certificate review procedures
  • Equipment performance: Establish equipment qualification and calibration schedules, document HPLC system suitability, validate synthesizer performance
  • Operator training: Develop standard operating procedures with visual aids, implement competency assessment, reduce operator-dependent steps through automation
  • Process control: Establish in-process testing checkpoints, implement statistical process control with control charts, define action limits triggering investigation

Process optimization employs quality-by-design (QbD) approaches including design of experiments (DOE) to evaluate multi-factor interactions, risk assessment to prioritize critical parameters, and statistical analysis to establish design space. Analytical method development and validation ensure accurate, precise measurement of quality attributes supporting optimization and manufacturing decisions. Technology transfer from R&D to manufacturing scale incorporates scale-up studies addressing mixing, heat transfer, and equipment-specific factors affecting product quality.

Conclusion

Successful manufacture of pharmaceutical-grade Melanotan-2 demands integration of synthetic chemistry expertise, analytical method development, quality systems implementation, and regulatory compliance strategies throughout the product lifecycle. The complexity of this cyclic heptapeptide, incorporating non-natural amino acids, lactam bridge formation, and multiple oxidation-sensitive residues, requires careful attention to synthesis conditions, purification methodology selection, and stability-protecting formulation approaches.

Manufacturing operations must balance quality objectives with economic considerations, optimizing crude synthesis purity to reduce purification burden while maintaining acceptable yields and minimizing cycle time. Process analytical technology and automation enable consistent quality while reducing operator-dependent variability. Comprehensive analytical characterization provides quality assurance and supports continuous improvement through trending and root cause analysis of deviations.

The specifications, protocols, and quality control strategies presented in this manufacturing profile represent industry best practices adapted from peptide pharmaceutical development. Manufacturers should customize these guidelines based on specific equipment capabilities, regulatory requirements for intended markets, and product applications while maintaining the fundamental quality principles of identity, purity, potency, and safety. Continued advancement in solid-phase synthesis chemistry, purification technology, and analytical instrumentation will further enhance manufacturing efficiency and product quality for this important melanocortin receptor agonist.

References and Resources

  1. Bednarek MA, et al. "Structure-function studies on the cyclic peptide MT-II, lactam derivative of alpha-melanotropin." Journal of Medicinal Chemistry. 1994;37(11):1558-1569. https://pubs.acs.org/doi/10.1021/jm00037a009
  2. International Conference on Harmonisation (ICH). "ICH Q7: Good Manufacturing Practice Guide for Active Pharmaceutical Ingredients." November 2000. https://www.ich.org/page/quality-guidelines
  3. Chan WC, White PD. Fmoc Solid Phase Peptide Synthesis: A Practical Approach. Oxford University Press; 2000. https://global.oup.com/academic/product/fmoc-solid-phase-peptide-synthesis-9780199637256
  4. Albericio F, Kruger HG. "Therapeutic peptides." Future Medicinal Chemistry. 2012;4(12):1527-1531. https://www.future-science.com/doi/10.4155/fmc.12.94
  5. ICH Harmonised Guideline. "Stability testing of new drug substances and products Q1A(R2)." February 2003. https://database.ich.org/sites/default/files/Q1A%28R2%29%20Guideline.pdf
  6. Palian MM, et al. "Analysis of the covalent structure of peptides and proteins." Current Protocols in Protein Science. 2016;Chapter 11:Unit 11.1.1-11.1.56. https://currentprotocols.onlinelibrary.wiley.com/doi/10.1002/0471140864.ps1101s84
  7. Henninot A, et al. "The Current State of Peptide Drug Discovery: Back to the Future?" Journal of Medicinal Chemistry. 2018;61(4):1382-1414. https://pubs.acs.org/doi/10.1021/acs.jmedchem.7b00318
  8. Valeur E, Bradley M. "Amide bond formation: beyond the myth of coupling reagents." Chemical Society Reviews. 2009;38(2):606-631. https://pubs.rsc.org/en/content/articlelanding/2009/cs/b701677h
  9. U.S. Food and Drug Administration. "Guidance for Industry: Q3C — Tables and List." Center for Drug Evaluation and Research (CDER), November 2017. https://www.fda.gov/regulatory-information/search-fda-guidance-documents/q3c-tables-and-list-guidance-industry
  10. Banga AK, editor. Therapeutic Peptides and Proteins: Formulation, Processing, and Delivery Systems. 3rd ed. CRC Press; 2015. https://www.taylorfrancis.com/books/mono/10.1201/b18392/therapeutic-peptides-proteins-ashim-banga